Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Med ; 22(7): 792-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27322744

ABSTRACT

Genetic heterogeneity contributes to clinical outcome and progression of most tumors, but little is known about allelic diversity for epigenetic compartments, and almost no data exist for acute myeloid leukemia (AML). We examined epigenetic heterogeneity as assessed by cytosine methylation within defined genomic loci with four CpGs (epialleles), somatic mutations, and transcriptomes of AML patient samples at serial time points. We observed that epigenetic allele burden is linked to inferior outcome and varies considerably during disease progression. Epigenetic and genetic allelic burden and patterning followed different patterns and kinetics during disease progression. We observed a subset of AMLs with high epiallele and low somatic mutation burden at diagnosis, a subset with high somatic mutation and lower epiallele burdens at diagnosis, and a subset with a mixed profile, suggesting distinct modes of tumor heterogeneity. Genes linked to promoter-associated epiallele shifts during tumor progression showed increased single-cell transcriptional variance and differential expression, suggesting functional impact on gene regulation. Thus, genetic and epigenetic heterogeneity can occur with distinct kinetics likely to affect the biological and clinical features of tumors.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Genetic Heterogeneity , Leukemia, Myeloid, Acute/genetics , Adult , Alleles , CpG Islands , Cytosine/metabolism , DNA Methylation , Disease Progression , Evolution, Molecular , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Multivariate Analysis , Prognosis , Promoter Regions, Genetic , Proportional Hazards Models , Sequence Analysis, DNA , Sequence Analysis, RNA , Survival Rate
3.
Elife ; 42015 Nov 17.
Article in English | MEDLINE | ID: mdl-26575292

ABSTRACT

RBM15, an RNA binding protein, determines cell-fate specification of many tissues including blood. We demonstrate that RBM15 is methylated by protein arginine methyltransferase 1 (PRMT1) at residue R578, leading to its degradation via ubiquitylation by an E3 ligase (CNOT4). Overexpression of PRMT1 in acute megakaryocytic leukemia cell lines blocks megakaryocyte terminal differentiation by downregulation of RBM15 protein level. Restoring RBM15 protein level rescues megakaryocyte terminal differentiation blocked by PRMT1 overexpression. At the molecular level, RBM15 binds to pre-messenger RNA intronic regions of genes important for megakaryopoiesis such as GATA1, RUNX1, TAL1 and c-MPL. Furthermore, preferential binding of RBM15 to specific intronic regions recruits the splicing factor SF3B1 to the same sites for alternative splicing. Therefore, PRMT1 regulates alternative RNA splicing via reducing RBM15 protein concentration. Targeting PRMT1 may be a curative therapy to restore megakaryocyte differentiation for acute megakaryocytic leukemia.


Subject(s)
Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Cell Line , Humans , Methylation , Proteolysis , Ubiquitination
4.
Nat Med ; 21(11): 1344-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437366

ABSTRACT

The tumor suppressors BAP1 and ASXL1 interact to form a polycomb deubiquitinase complex that removes monoubiquitin from histone H2A lysine 119 (H2AK119Ub). However, BAP1 and ASXL1 are mutated in distinct cancer types, consistent with independent roles in regulating epigenetic state and malignant transformation. Here we demonstrate that Bap1 loss in mice results in increased trimethylated histone H3 lysine 27 (H3K27me3), elevated enhancer of zeste 2 polycomb repressive complex 2 subunit (Ezh2) expression, and enhanced repression of polycomb repressive complex 2 (PRC2) targets. These findings contrast with the reduction in H3K27me3 levels seen with Asxl1 loss. Conditional deletion of Bap1 and Ezh2 in vivo abrogates the myeloid progenitor expansion induced by Bap1 loss alone. Loss of BAP1 results in a marked decrease in H4K20 monomethylation (H4K20me1). Consistent with a role for H4K20me1 in the transcriptional regulation of EZH2, expression of SETD8-the H4K20me1 methyltransferase-reduces EZH2 expression and abrogates the proliferation of BAP1-mutant cells. Furthermore, mesothelioma cells that lack BAP1 are sensitive to EZH2 pharmacologic inhibition, suggesting a novel therapeutic approach for BAP1-mutant malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histones/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Knockout Techniques , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunoprecipitation , Leukemia/genetics , Mesothelioma/genetics , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
5.
Cancer Cell ; 27(4): 502-15, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25873173

ABSTRACT

Specific combinations of acute myeloid leukemia (AML) disease alleles, including FLT3 and TET2 mutations, confer distinct biologic features and adverse outcome. We generated mice with mutations in Tet2 and Flt3, which resulted in fully penetrant, lethal AML. Multipotent Tet2(-/-);Flt3(ITD) progenitors (LSK CD48(+)CD150(-)) propagate disease in secondary recipients and were refractory to standard AML chemotherapy and FLT3-targeted therapy. Flt3(ITD) mutations and Tet2 loss cooperatively remodeled DNA methylation and gene expression to an extent not seen with either mutant allele alone, including at the Gata2 locus. Re-expression of Gata2 induced differentiation in AML stem cells and attenuated leukemogenesis. TET2 and FLT3 mutations cooperatively induce AML, with a defined leukemia stem cell population characterized by site-specific changes in DNA methylation and gene expression.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics , Antineoplastic Agents/therapeutic use , Cell Differentiation/genetics , Cytarabine/therapeutic use , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Doxorubicin/therapeutic use , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Haploinsufficiency , Mutation , Proto-Oncogene Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism
6.
Stem Cell Reports ; 4(4): 658-69, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25754204

ABSTRACT

Epigenetic regulation of key transcriptional programs is a critical mechanism that controls hematopoietic development, and, thus, aberrant expression patterns or mutations in epigenetic regulators occur frequently in hematologic malignancies. We demonstrate that the Polycomb protein L3MBTL1, which is monoallelically deleted in 20q- myeloid malignancies, represses the ability of stem cells to drive hematopoietic-specific transcriptional programs by regulating the expression of SMAD5 and impairing its recruitment to target regulatory regions. Indeed, knockdown of L3MBTL1 promotes the development of hematopoiesis and impairs neural cell fate in human pluripotent stem cells. We also found a role for L3MBTL1 in regulating SMAD5 target gene expression in mature hematopoietic cell populations, thereby affecting erythroid differentiation. Taken together, we have identified epigenetic priming of hematopoietic-specific transcriptional networks, which may assist in the development of therapeutic approaches for patients with anemia.


Subject(s)
Cell Differentiation/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Smad5 Protein/metabolism , Transcription, Genetic , Down-Regulation , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Repressor Proteins , Smad5 Protein/genetics , Tumor Suppressor Proteins
7.
Cancer Discov ; 5(3): 316-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572172

ABSTRACT

UNLABELLED: The identification of JAK2/MPL mutations in patients with myeloproliferative neoplasms (MPN) has led to the clinical development of JAK kinase inhibitors, including ruxolitinib. Ruxolitinib reduces splenomegaly and systemic symptoms in myelofibrosis and improves overall survival; however, the mechanism by which JAK inhibitors achieve efficacy has not been delineated. Patients with MPN present with increased levels of circulating proinflammatory cytokines, which are mitigated by JAK inhibitor therapy. We sought to elucidate mechanisms by which JAK inhibitors attenuate cytokine-mediated pathophysiology. Single-cell profiling demonstrated that hematopoietic cells from myelofibrosis models and patient samples aberrantly secrete inflammatory cytokines. Pan-hematopoietic Stat3 deletion reduced disease severity and attenuated cytokine secretion, with similar efficacy as observed with ruxolitinib therapy. In contrast, Stat3 deletion restricted to MPN cells did not reduce disease severity or cytokine production. Consistent with these observations, we found that malignant and nonmalignant cells aberrantly secrete cytokines and JAK inhibition reduces cytokine production from both populations. SIGNIFICANCE: Our results demonstrate that JAK-STAT3-mediated cytokine production from malignant and nonmalignant cells contributes to MPN pathogenesis and that JAK inhibition in both populations is required for therapeutic efficacy. These findings provide novel insight into the mechanisms by which JAK kinase inhibition achieves therapeutic efficacy in MPNs.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Janus Kinases/metabolism , Myeloproliferative Disorders/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cytokines/metabolism , Disease Models, Animal , Gene Deletion , Humans , Inflammation Mediators/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinases/genetics , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Knockout , Mutation , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Protein Kinase Inhibitors/pharmacology , STAT Transcription Factors/genetics , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 111(50): E5401-10, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25516983

ABSTRACT

Patients with myeloproliferative neoplasms (MPNs) are at significant, cumulative risk of leukemic transformation to acute myeloid leukemia (AML), which is associated with adverse clinical outcome and resistance to standard AML therapies. We performed genomic profiling of post-MPN AML samples; these studies demonstrate somatic tumor protein 53 (TP53) mutations are common in JAK2V617F-mutant, post-MPN AML but not in chronic-phase MPN and lead to clonal dominance of JAK2V617F/TP53-mutant leukemic cells. Consistent with these data, expression of JAK2V617F combined with Tp53 loss led to fully penetrant AML in vivo. JAK2V617F-mutant, Tp53-deficient AML was characterized by an expanded megakaryocyte erythroid progenitor population that was able to propagate the disease in secondary recipients. In vitro studies revealed that post-MPN AML cells were sensitive to decitabine, the JAK1/2 inhibitor ruxolitinib, or the heat shock protein 90 inhibitor 8-(6-iodobenzo[d][1.3]dioxol-5-ylthio)-9-(3-(isopropylamino)propyl)-9H-purine-6-amine (PU-H71). Treatment with ruxolitinib or PU-H71 improved survival of mice engrafted with JAK2V617F-mutant, Tp53-deficient AML, demonstrating therapeutic efficacy for these targeted therapies and providing a rationale for testing these therapies in post-MPN AML.


Subject(s)
Hematologic Neoplasms/complications , Janus Kinase 2/genetics , Leukemia, Myeloid, Acute/genetics , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Tumor Suppressor Protein p53/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Benzodioxoles/pharmacology , Blotting, Western , Colony-Forming Units Assay , Decitabine , Exome/genetics , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/etiology , Mice , Mutation, Missense/genetics , Nitriles , Purines/pharmacology , Pyrazoles/pharmacology , Pyrimidines
9.
Cell Rep ; 9(5): 1841-1855, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25482556

ABSTRACT

Somatic mutations in IDH1/IDH2 and TET2 result in impaired TET2-mediated conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). The observation that WT1 inactivating mutations anticorrelate with TET2/IDH1/IDH2 mutations in acute myeloid leukemia (AML) led us to hypothesize that WT1 mutations may impact TET2 function. WT1 mutant AML patients have reduced 5hmC levels similar to TET2/IDH1/IDH2 mutant AML. These mutations are characterized by convergent, site-specific alterations in DNA hydroxymethylation, which drive differential gene expression more than alterations in DNA promoter methylation. WT1 overexpression increases global levels of 5hmC, and WT1 silencing reduced 5hmC levels. WT1 physically interacts with TET2 and TET3, and WT1 loss of function results in a similar hematopoietic differentiation phenotype as observed with TET2 deficiency. These data provide a role for WT1 in regulating DNA hydroxymethylation and suggest that TET2 IDH1/IDH2 and WT1 mutations define an AML subtype defined by dysregulated DNA hydroxymethylation.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/physiology , WT1 Proteins/genetics , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Cytosine/analogs & derivatives , Cytosine/physiology , Dioxygenases , Enhancer Elements, Genetic , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, Knockout , Mutation , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA
10.
Blood ; 123(22): e123-33, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24740812

ABSTRACT

Genomic studies have identified somatic alterations in the majority of myeloproliferative neoplasms (MPN) patients, including JAK2 mutations in the majority of MPN patients and CALR mutations in JAK2-negative MPN patients. However, the role of JAK-STAT pathway activation in different MPNs, and in patients without JAK2 mutations, has not been definitively delineated. We used expression profiling, single nucleotide polymorphism arrays, and mutational profiling to investigate a well-characterized cohort of MPN patients. MPN patients with homozygous JAK2V617F mutations were characterized by a distinctive transcriptional profile. Notably, a transcriptional signature consistent with activated JAK2 signaling is seen in all MPN patients regardless of clinical phenotype or mutational status. In addition, the activated JAK2 signature was present in patients with somatic CALR mutations. Conversely, we identified a gene expression signature of CALR mutations; this signature was significantly enriched in JAK2-mutant MPN patients consistent with a shared mechanism of transformation by JAK2 and CALR mutations. We also identified a transcriptional signature of TET2 mutations in MPN patent samples. Our data indicate that MPN patients, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of the JAK-STAT pathway in MPN pathogenesis.


Subject(s)
Genomics , Janus Kinases/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Calreticulin , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Female , Gene Expression Profiling , Homozygote , Humans , Janus Kinase 2/genetics , Janus Kinases/genetics , Male , Mutation , STAT Transcription Factors/genetics , Transcriptome
11.
J Leuk (Los Angel) ; 2(3)2014 Sep.
Article in English | MEDLINE | ID: mdl-28239612

ABSTRACT

Acute Myeloid Leukemia (AML) is a highly heterogeneous and poor prognosis disease with few available therapeutic options. Novel advances are urgently needed, however effective models to test experimental therapeutics have been lacking. Recently, NOD/SCID/IL2rγnull (NSG) mice were shown to engraft primary human AML in a manner that recapitulated the natural disease and its progression. Additionally, integrated genomic profiling was used to refine risk stratification of AML. In this study, we demonstrated the engraftment of molecularly defined primary AML in NSG mice. We showed that AML that express DNMT3A mutations, which predict for adverse outcome, engrafted with exceptional efficacy. Lastly, we demonstrated that human AML-engrafted NSG mice can be effectively used to study novel ceramide-based therapeutics. Ceramide is a bioactive sphingolipid that has been implicated as an inducer of apoptosis. Elevation in cancer cell ceramide levels either via exogenous delivery or by provoking intracellular ceramide generation is the goal of ceramide-based therapeutics. In this study, we used the human AML-engrafted NSG mouse model to evaluate nanoliposomal short-chain C6-ceramide and a nanoliposomal formulation of the ceramide-inducer tamoxifen. Altogether, the NSG model is likely to prove invaluable in the study of novel agents, sushc as ceramide-based therapeutics, with the ability to define therapeutic activity against specific molecularly defined and risk stratified AML.

12.
J Exp Med ; 210(12): 2641-59, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24218140

ABSTRACT

Somatic Addition of Sex Combs Like 1 (ASXL1) mutations occur in 10-30% of patients with myeloid malignancies, most commonly in myelodysplastic syndromes (MDSs), and are associated with adverse outcome. Germline ASXL1 mutations occur in patients with Bohring-Opitz syndrome. Here, we show that constitutive loss of Asxl1 results in developmental abnormalities, including anophthalmia, microcephaly, cleft palates, and mandibular malformations. In contrast, hematopoietic-specific deletion of Asxl1 results in progressive, multilineage cytopenias and dysplasia in the context of increased numbers of hematopoietic stem/progenitor cells, characteristic features of human MDS. Serial transplantation of Asxl1-null hematopoietic cells results in a lethal myeloid disorder at a shorter latency than primary Asxl1 knockout (KO) mice. Asxl1 deletion reduces hematopoietic stem cell self-renewal, which is restored by concomitant deletion of Tet2, a gene commonly co-mutated with ASXL1 in MDS patients. Moreover, compound Asxl1/Tet2 deletion results in an MDS phenotype with hastened death compared with single-gene KO mice. Asxl1 loss results in a global reduction of H3K27 trimethylation and dysregulated expression of known regulators of hematopoiesis. RNA-Seq/ChIP-Seq analyses of Asxl1 in hematopoietic cells identify a subset of differentially expressed genes as direct targets of Asxl1. These findings underscore the importance of Asxl1 in Polycomb group function, development, and hematopoiesis.


Subject(s)
Abnormalities, Multiple/etiology , Myelodysplastic Syndromes/etiology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Abnormalities, Multiple/genetics , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Deletion , Germ-Line Mutation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phenotype , Pregnancy , Protein Binding , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism
13.
Am J Hematol ; 88(9): 723-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23720412

ABSTRACT

Erythropoiesis is a tightly regulated process which becomes decoupled from its normal differentiation program in patients with polycythemia vera (PV). Somatic mutations in JAK2 are commonly associated with this myeloid proliferative disorder. To gain insight into the molecular events that are required for abnormally developing erythroid cells to escape dependence on normal growth signals, we performed in vitro expansion of mature erythroblasts (ERY) from seven normal healthy donors and from seven polycythemic patients in the presence of IL3, EPO, SCF for 10, 11, or 13 days. Normal ERYs required exposure to the glucocorticoid dexamethasone (Dex) for expansion, while PV-derived ERYs expanded in the absence of dexamethasone. RNA expression profiling revealed enrichment of two known oncogenes, GPR56 and RAB4a, in PV-derived ERYs along with reduced expression levels of transcription factor TAL1 (ANOVA FDR < 0.05). While both normal and polycythemic-derived ERYs integrated signaling cascades for growth, they did so via different signaling pathways which are represented by their differential phospho-profiles. Our results show that normal ERYs displayed greater levels of phosphorylation of EGFR, PDGFRß, TGFß, and cKit, while PV-derived ERYs were characterized by increased phosphorylation of cytoplasmic kinases in the JAK/STAT, PI3K, and GATA1 pathways. Together these data suggest that PV erythroblast expansion and maturation may be maintained and enriched in the absence of dexamethasone through reduced TAL1 expression and by accessing additional signaling cascades. Members of this acquired repertoire may provide important insight into the pathogenesis of aberrant erythropoiesis in myeloproliferative neoplasms such as polycythemia vera.


Subject(s)
Erythroblasts/metabolism , Erythropoiesis/genetics , Phosphoproteins/genetics , Polycythemia Vera/genetics , Adult , Aged , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Case-Control Studies , Cells, Cultured , Dexamethasone/pharmacology , Erythroblasts/drug effects , Erythroblasts/pathology , Erythropoietin/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-3/pharmacology , Male , Middle Aged , Phosphoproteins/metabolism , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Proteomics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cell Factor/pharmacology , T-Cell Acute Lymphocytic Leukemia Protein 1 , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
14.
Cancer Cell ; 22(2): 180-93, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22897849

ABSTRACT

Recurrent somatic ASXL1 mutations occur in patients with myelodysplastic syndrome, myeloproliferative neoplasms, and acute myeloid leukemia, and are associated with adverse outcome. Despite the genetic and clinical data implicating ASXL1 mutations in myeloid malignancies, the mechanisms of transformation by ASXL1 mutations are not understood. Here, we identify that ASXL1 mutations result in loss of polycomb repressive complex 2 (PRC2)-mediated histone H3 lysine 27 (H3K27) tri-methylation. Through integration of microarray data with genome-wide histone modification ChIP-Seq data, we identify targets of ASXL1 repression, including the posterior HOXA cluster that is known to contribute to myeloid transformation. We demonstrate that ASXL1 associates with the PRC2, and that loss of ASXL1 in vivo collaborates with NRASG12D to promote myeloid leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Mutation/genetics , Myeloid Cells/pathology , Repressor Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Hematopoietic System/metabolism , Hematopoietic System/pathology , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Methylation , Mice , Myeloid Cells/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Binding , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Up-Regulation/genetics , ras Proteins/metabolism
15.
Nature ; 489(7414): 155-9, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22820254

ABSTRACT

The identification of somatic activating mutations in JAK2 (refs 1­4) and in the thrombopoietin receptor gene (MPL) in most patients with myeloproliferative neoplasm (MPN) led to the clinical development of JAK2 kinase inhibitors. JAK2 inhibitor therapy improves MPN-associated splenomegaly and systemic symptoms but does not significantly decrease or eliminate the MPN clone in most patients with MPN. We therefore sought to characterize mechanisms by which MPN cells persist despite chronic inhibition of JAK2. Here we show that JAK2 inhibitor persistence is associated with reactivation of JAK­STAT signalling and with heterodimerization between activated JAK2 and JAK1 or TYK2, consistent with activation of JAK2 in trans by other JAK kinases. Further, this phenomenon is reversible: JAK2 inhibitor withdrawal is associated with resensitization to JAK2 kinase inhibitors and with reversible changes in JAK2 expression. We saw increased JAK2 heterodimerization and sustained JAK2 activation in cell lines, in murine models and in patients treated with JAK2 inhibitors. RNA interference and pharmacological studies show that JAK2-inhibitor-persistent cells remain dependent on JAK2 protein expression. Consequently, therapies that result in JAK2 degradation retain efficacy in persistent cells and may provide additional benefit to patients with JAK2-dependent malignancies treated with JAK2 inhibitors.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Myeloproliferative Disorders/drug therapy , Protein Multimerization , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Cell Line , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Granulocytes/drug effects , Granulocytes/enzymology , Granulocytes/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Janus Kinase 1/biosynthesis , Janus Kinase 1/deficiency , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Mice , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phosphorylation , Protein Biosynthesis , RNA Interference , Signal Transduction/drug effects , TYK2 Kinase/biosynthesis , TYK2 Kinase/deficiency , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
16.
Blood ; 119(19): 4480-5, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22431577

ABSTRACT

Leukemic transformation (LT) of myeloproliferative neoplasms (MPNs) is associated with a poor prognosis and resistance to therapy. Although previous candidate genetic studies have identified mutations in MPN patients who develop acute leukemia, the complement of genetic abnormalities in MPN patients who undergo LT is not known nor have specific molecular abnormalities been shown to have clinical relevance in this setting. We performed high-throughput resequencing of 22 genes in 53 patients with LT after MPN to characterize the frequency of known myeloid mutations in this entity. In addition to JAK2 and TET2 mutations, which occur commonly in LT after MPN, we identified recurrent mutations in the serine/arginine-rich splicing factor 2 (SRSF2) gene (18.9%) in acute myeloid leukemia (AML) transformed from MPNs. SRSF2 mutations are more common in AML derived from MPNs compared with LT after myelodysplasia (4.8%) or de novo AML (5.6%), respectively (P=.05). Importantly, SRSF2 mutations are associated with worsened overall survival in MPN patients who undergo LT in univariate (P=.03; HR, 2.77; 95% CI, 1.10-7.00) and multivariate analysis (P<.05; HR, 2.11; 95% CI, 1.01-4.42). These data suggest that SRSF2 mutations contribute to the pathogenesis of LT and may guide novel therapeutic approaches for MPN patients who undergo LT.


Subject(s)
Cell Transformation, Neoplastic/genetics , Hematologic Neoplasms/genetics , Leukemia/pathology , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Nuclear Proteins/genetics , Ribonucleoproteins/genetics , Base Sequence , Cell Transformation, Neoplastic/pathology , Cohort Studies , DNA Mutational Analysis , Disease Progression , Gene Frequency , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Humans , Leukemia/diagnosis , Leukemia/genetics , Leukemia/mortality , Mutation/physiology , Myeloproliferative Disorders/mortality , Myeloproliferative Disorders/pathology , Prognosis , Serine-Arginine Splicing Factors , Spliceosomes/genetics , Spliceosomes/metabolism
17.
Clin Transl Oncol ; 13(3): 194-203, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21421465

ABSTRACT

OBJECTIVE: Pax3 and Pax7 are closely related genes that are involved in commitment of cells to a myogenic lineage during skeletal muscle development and regeneration. Several Pax3 and Pax7 transcripts are expressed from the genes, generating different isoforms with potentially distinct DNA binding and transactivation properties. The aim of this study was to investigate the implication of Pax3 and Pax7 C-terminal isoforms during myogenic differentiation and tumorigenesis, since fusions involving these genes are commonly associated with alveolar rhabdomyosarcoma (ARMS). METHODS: Uncommitted (mouse mesenchymal stem cells, MSCs) and committed (C2C12) myogenic precursor cells were stably transfected with PAX3/FKHR and PAXC7/ FKHR fusion genes. We analysed gene and protein expression comparing the newly generated cells with the parental cells, to determine the functional importance of Pax3 and Pax7 C-terminal isoforms. RESULTS: We found that the transcript Pax3c was expressed at low levels in undifferentiated C2C12 and MSCs cells, but its expression levels increased considerably at later stages of differentiation. However, expression levels of Pax3d transcript increased only slightly after differentiation. Pax7 transcripts, present before differentiation in committed C2C12 cells, but absent in uncommitted MSCs, increased noticeably in MSCs after differentiation. We also found that the presence of PAX/FKHR fusions prevented both C2C12 and MSC cells from terminal myogenic differentiation and increased the expression of discrete endogenous Pax3/7 transcripts, in particular Pax3d and Pax7B. CONCLUSIONS: Our results suggest that both Pax3 and Pax7 transcripts are required for commitment of cells to the myogenic lineage, with each transcript having a distinct role. More specifically, the Pax3c isoform may be required for terminal myogenic differentiation whereas the Pax3d isoform may be involved in undifferentiated cell maintenance and/or proliferation.


Subject(s)
Cell Differentiation/physiology , Muscle Cells/cytology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Animals , Cell Line , Cell Lineage/physiology , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mice , Muscle Cells/metabolism , PAX3 Transcription Factor , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma, Alveolar/metabolism , Transfection
18.
Mol Cancer Ther ; 10(2): 313-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21216937

ABSTRACT

To formally explore the potential therapeutic effect of histone deacetylase inhibitors (HDACI) and DNA-methyltransferase inhibitors (DNA-MI) on sarcomas, we treated a large sarcoma cell line panel with five different HDACIs in the absence and presence of the DNA-MI decitabine. We observed that the IC(50) value of each HDACI was consistent for all cell lines whereas decitabine as a single agent showed no growth inhibition at standard doses. Combination HDACI/DNA-MI therapy showed a preferential synergism for specific sarcoma cell lines. Subsequently, we identified and validated (in vitro and in vivo) a two-gene set signature (high CUGBP2; low RHOJ) that associated with the synergistic phenotype. We further uncover that the epigenetic synergism leading to specific upregulation of CDKI p21 in specific cell lines is a function of the differences in the degree of baseline chromatin modification. Finally, we show that these chromatin and gene expression patterns are similarly present in the majority of high-grade primary sarcomas. Our results provide the first demonstration of a gene set that can predict responsiveness to HDACI/DNA-MI and links this responsiveness mechanistically to the baseline chromatin structure.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Enzyme Inhibitors/pharmacology , Epigenomics , Histone Deacetylase Inhibitors/pharmacology , Sarcoma/genetics , Sarcoma/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cluster Analysis , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, SCID , Random Allocation , Xenograft Model Antitumor Assays
19.
Hum Cell ; 22(4): 85-93, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19874397

ABSTRACT

To expand the available tools for investigating human sarcomas, we characterized the primary properties of 22 common, uncommon, and newly characterized sarcoma cell lines representing eight different histological subtypes. Throughout the characterization process we noticed that in vitro markers and assays are poor indicators of tumorigenicity and that generated xenografts often bear little resemblance to the original histopathology. In vitro properties examined included morphology, proliferation rate, cell cycle characteristics, invasiveness, and immunohistochemical expression of p53 and phospho-AKT. In vivo properties examined included days to tumor formation in NOD/SCID mice, xenograft morphology in several locations and immunohistochemical expression of Ki67, p53 and phospho-AKT. We believe that such an in depth comparison of a large cohort of sarcoma cell lines will be useful in both designing and interpreting experiments aimed at elucidating both the molecular biology and efficacy of therapeutic agents in sarcomas. However, that data generated also suggests a small set of sarcoma cell lines may be inappropriate for generalizations regarding biological behavior of specific sarcoma subtypes. Integration of functional genomics or other more sophisticated assays of cell lines may help bridge the differences in vitro and in vivo.


Subject(s)
Sarcoma/pathology , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Humans , In Vitro Techniques , Ki-67 Antigen/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/metabolism , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
20.
Expert Rev Anticancer Ther ; 9(8): 1135-44, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671033

ABSTRACT

The essence and origin of malignant fibrous histiocytoma (MFH) have been debated for now close to five decades. Originally characterized as a morphologically unique soft-tissue sarcoma subtype of unclear etiology in 1963, with a following 15 years of research only to conclude that "the issue of histogenesis [of MFH] is largely unresolvable"; it is "now regarded as synonymous with [high grade] undifferentiated pleomorphic sarcoma and essentially represents a diagnosis of exclusion". Yet despite this apparent lack of progress, the first decade of the 21st century has seen some significant progress in terms of defining the origins of MFH. Perhaps more importantly these origins might also pave the way for novel therapies. This manuscript will highlight MFH's troubled history, discuss recent advances, and comment as to what the coming years may promise and what further needs to be done to make sure that progress continues.


Subject(s)
Histiocytoma, Malignant Fibrous/diagnosis , Mesenchymal Stem Cells/metabolism , Sarcoma/diagnosis , Animals , Diagnosis, Differential , Histiocytoma, Malignant Fibrous/classification , Histiocytoma, Malignant Fibrous/history , History, 21st Century , Humans , Sarcoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...